Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.714
Filtrar
1.
CNS Neurosci Ther ; 30(6): e14794, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38867379

RESUMO

BACKGROUND: Radiation-induced brain injury is a neurological condition resulting from radiotherapy for malignant tumors, with its underlying pathogenesis still not fully understood. Current hypotheses suggest that immune cells, particularly the excessive activation of microglia in the central nervous system and the migration of peripheral immune cells into the brain, play a critical role in initiating and progressing the injury. This review aimed to summarize the latest advances in the cellular and molecular mechanisms and the therapeutic potential of microglia in radiation-induced brain injury. METHODS: This article critically examines recent developments in understanding the role of microglia activation in radiation-induced brain injury. It elucidates associated mechanisms and explores novel research pathways and therapeutic options for managing this condition. RESULTS: Post-irradiation, activated microglia release numerous inflammatory factors, exacerbating neuroinflammation and facilitating the onset and progression of radiation-induced damage. Therefore, controlling microglial activation and suppressing the secretion of related inflammatory factors is crucial for preventing radiation-induced brain injury. While microglial activation is a primary factor in neuroinflammation, the precise mechanisms by which radiation prompts this activation remain elusive. Multiple signaling pathways likely contribute to microglial activation and the progression of radiation-induced brain injury. CONCLUSIONS: The intricate microenvironment and molecular mechanisms associated with radiation-induced brain injury underscore the crucial roles of immune cells in its onset and progression. By investigating the interplay among microglia, neurons, astrocytes, and peripheral immune cells, potential strategies emerge to mitigate microglial activation, reduce the release of inflammatory agents, and impede the entry of peripheral immune cells into the brain.


Assuntos
Lesões Encefálicas , Microglia , Lesões por Radiação , Microglia/efeitos da radiação , Microglia/metabolismo , Animais , Humanos , Lesões por Radiação/metabolismo , Lesões por Radiação/terapia , Lesões Encefálicas/etiologia , Lesões Encefálicas/metabolismo , Doenças Neuroinflamatórias/etiologia
2.
Cells ; 13(11)2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38891060

RESUMO

Mitochondrial aldehyde dehydrogenase-2 (ALDH2) metabolizes acetaldehyde to acetate. People with ALDH2 deficiency and Aldh2-knockout (KO) mice are more susceptible to alcohol-induced tissue damage. However, the underlying mechanisms behind ALDH2-related gut-associated brain damage remain unclear. Age-matched young female Aldh2-KO and C57BL/6J wild-type (WT) mice were gavaged with binge alcohol (4 g/kg/dose, three doses) or dextrose (control) at 12 h intervals. Tissues and sera were collected 1 h after the last ethanol dose and evaluated by histological and biochemical analyses of the gut and hippocampus and their extracts. For the mechanistic study, mouse neuroblast Neuro2A cells were exposed to ethanol with or without an Aldh2 inhibitor (Daidzin). Binge alcohol decreased intestinal tight/adherens junction proteins but increased oxidative stress-mediated post-translational modifications (PTMs) and enterocyte apoptosis, leading to elevated gut leakiness and endotoxemia in Aldh2-KO mice compared to corresponding WT mice. Alcohol-exposed Aldh2-KO mice also showed higher levels of hippocampal brain injury, oxidative stress-related PTMs, and neuronal apoptosis than the WT mice. Additionally, alcohol exposure reduced Neuro2A cell viability with elevated oxidative stress-related PTMs and apoptosis, all of which were exacerbated by Aldh2 inhibition. Our results show for the first time that ALDH2 plays a protective role in binge alcohol-induced brain injury partly through the gut-brain axis, suggesting that ALDH2 is a potential target for attenuating alcohol-induced tissue injury.


Assuntos
Aldeído-Desidrogenase Mitocondrial , Consumo Excessivo de Bebidas Alcoólicas , Lesões Encefálicas , Camundongos Endogâmicos C57BL , Camundongos Knockout , Animais , Aldeído-Desidrogenase Mitocondrial/metabolismo , Aldeído-Desidrogenase Mitocondrial/genética , Camundongos , Consumo Excessivo de Bebidas Alcoólicas/patologia , Lesões Encefálicas/patologia , Lesões Encefálicas/metabolismo , Etanol/toxicidade , Etanol/farmacologia , Feminino , Apoptose/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Hipocampo/patologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos
3.
J Cell Mol Med ; 28(11): e18366, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38856956

RESUMO

Ischemic stroke is one of the main causes of disability and death. However, recanalization of occluded cerebral arteries is effective only within a very narrow time window. Therefore, it is particularly important to find neuroprotective biological targets for cerebral artery recanalization. Here, gene expression profiles of datasets GSE160500 and GSE97537 were downloaded from the GEO database, which were related to ischemic stroke in rats. Olfactory receptor 78 (Olfr78) was screened, and which highly associated with Calcium signalling pathway and MAPK pathway. Interacting protein of Olfr78, Prkaca, was predicted by STRING, and their interaction was validated by Co-IP analysis. Then, a rat model of middle cerebral artery occlusion/reperfusion (MCAO/R) and a neuronal cell model stimulated by oxygen-glucose deprivation/reoxygenation (OGD/R) were constructed, and the results showed that expression of Olfr78 and Prkaca was downregulated in MCAO rats and OGD/R-stimulated neurons. Overexpression of Olfr78 or Prkaca inhibited the secretion of inflammatory factors, Ca2+ overload, and OGD/R-induced neuronal apoptosis. Moreover, Overexpression of Prkaca increased protein levels of cAMP, PKA and phosphorylated p38 in OGD/R-stimulated neurons, while SB203580, a p38 inhibitor, treatment inhibited activation of the cAMP/PKA-MAPK pathway and counteracted the effect of Olfr78 overexpression on improvement of neuronal functions. Meanwhile, overexpression of Olfr78 or Prkaca markedly inhibited neuronal apoptosis and improved brain injury in MCAO/R rats. In conclusion, overexpression of Olfr78 inhibited Ca2+ overload and reduced neuronal apoptosis in MCAO/R rats by promoting Prkaca-mediated activation of the cAMP/PKA-MAPK pathway, thereby improving brain injury in cerebral ischaemia-reperfusion.


Assuntos
Apoptose , AMP Cíclico , Ratos Sprague-Dawley , Receptores Odorantes , Traumatismo por Reperfusão , Animais , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/genética , Ratos , Masculino , AMP Cíclico/metabolismo , Receptores Odorantes/metabolismo , Receptores Odorantes/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Lesões Encefálicas/metabolismo , Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Neurônios/metabolismo , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/metabolismo , Transdução de Sinais
4.
Neuropathol Appl Neurobiol ; 50(3): e12992, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38831600

RESUMO

PURPOSE: Radiation-induced brain injury, one of the side effects of cranial radiotherapy in tumour patients, usually results in durable and serious cognitive disorders. Microglia are important innate immune-effector cells in the central nervous system. However, the interaction between microglia and neurons in radiation-induced brain injury remains uncharacterised. METHODS AND MATERIALS: We established a microglia-neuron indirect co-culture model to assess the interaction between them. Microglia exposed to radiation were examined for pyroptosis using lactate dehydrogenase (LDH) release, Annexin V/PI staining, SYTOX staining and western blot. The role of nucleotide-binding oligomerisation domain-like receptor family pyrin domain containing 3 (NLRP3) was investigated in microglia exposed to radiation and in mouse radiation brain injury model through siRNA or inhibitor. Mini-mental state examination and cytokines in blood were performed in 23 patients who had experienced cranial irradiation. RESULTS: Microglia exerted neurotoxic features after radiation in the co-culture model. NLRP3 was up-regulated in microglia exposed to radiation, and then caspase-1 was activated. Thus, the gasdermin D protein was cleaved, and it triggered pyroptosis in microglia, which released inflammatory cytokines. Meanwhile, treatment with siRNA NLRP3 in vitro and NLRP3 inhibitor in vivo attenuated the damaged neuron cell and cognitive impairment, respectively. What is more, we found that the patients after radiation with higher IL-6 were observed to have a decreased MMSE score. CONCLUSIONS: These findings indicate that radiation-induced pyroptosis in microglia may promote radiation-induced brain injury via the secretion of neurotoxic cytokines. NLRP3 was evaluated as an important mediator in radiation-induced pyroptosis and a promising therapeutic target for radiation-induced brain injury.


Assuntos
Lesões Encefálicas , Microglia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Piroptose , Piroptose/efeitos da radiação , Piroptose/fisiologia , Microglia/metabolismo , Microglia/efeitos da radiação , Microglia/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Camundongos , Humanos , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Lesões Encefálicas/etiologia , Masculino , Neurônios/metabolismo , Neurônios/patologia , Neurônios/efeitos da radiação , Técnicas de Cocultura , Lesões por Radiação/patologia , Lesões por Radiação/metabolismo , Feminino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade
5.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 295-302, 2024 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-38814198

RESUMO

Aneurismal subarachnoid hemorrhage (aSAH) is a common disease in the neural system, with high death rate. Our study aimed to explore the clinical effect of external ventricular drainage under intracranial pressure monitoring in the treatment of patients with aSAH and investigate the role along with mechanism of miR-146a-5p in aSAH. Ninety-six aSAH patients were allocated into control group (CG) and study group (SG). The CG was released by lumbar puncture. The SG underwent external ventricular drainage based on intracranial pressure monitoring. The prognosis, daily living ability, neurological function, S100ß and NSE (neuron-specific enolase) levels and incidence of complications were monitored. Besides, a rat model of SAH was built to assess the neurobehavioral function, blood-brain barrier permeability, brain water content, neuronal apoptosis as well as inflammation. SAH cell model stimulated by oxyhemoglobin, and cell apoptosis as well as inflammation were measured. Luciferase reporter assay was implemented to explore the interaction between miR-146a-5p and STC1. Results showed higher GOS and BI scores but lower NIHSS scores, S100ß and NSE levels and complication rates in SG compared with CG. Additionally, miR-146a-5p presented down-regulation in brain tissues of SAH rat model, and overexpressed miR-146a-5p reduced brain injury along with neuroinflammation in SAH rat model. Oxyhemoglobin-induced nerve cell apoptosis along with inflammation after SAH, and overexpressed miR-146a-5p repressed oxyhemoglobin-induced nerve cell apoptosis along with inflammation. STC1 is the target mRNA of miR-146a-5p, and overexpressed miR-146a-5p represses oxyhemoglobin-induced nerve cell apoptosis along with inflammation via regulating STC1 expression. In conclusion, external ventricular drainage under intracranial pressure monitoring could promote prognosis, promote daily living ability, improve neurological function, reduce S100ß protein and NSE levels, and reduce the incidence of complications in patients with aSAH. Meanwhile, miR-146a-5p inhibited early brain injury and neuroinflammation in aSAH via regulating STC1 expression.


Assuntos
Apoptose , Lesões Encefálicas , Pressão Intracraniana , MicroRNAs , Hemorragia Subaracnóidea , MicroRNAs/genética , MicroRNAs/metabolismo , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/complicações , Animais , Humanos , Masculino , Lesões Encefálicas/etiologia , Lesões Encefálicas/metabolismo , Ratos , Pessoa de Meia-Idade , Feminino , Ratos Sprague-Dawley , Subunidade beta da Proteína Ligante de Cálcio S100/metabolismo , Subunidade beta da Proteína Ligante de Cálcio S100/genética , Drenagem/métodos , Modelos Animais de Doenças , Barreira Hematoencefálica/metabolismo , Fosfopiruvato Hidratase/metabolismo
6.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 220-225, 2024 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-38814212

RESUMO

This study explored the impact of penehyclidine hydrochloride on cognitive function in rats with brain injury. Sprague-Dawley rats (n=36) were randomly assigned to sham-operation, model, and penehyclidine hydrochloride groups. Rats in the sham-operation group underwent craniotomy, while the model and penehyclidine hydrochloride groups received brain injury models and interventions with normal saline and penehyclidine hydrochloride, respectively. Specimens were obtained two weeks post-intervention. Neurological deficits were evaluated using Zea-Longa scores, and memory was assessed with the Morris water maze test. ELISA determined brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) content. mRNA expressions of BDNF and NGF were assessed via qPCR, and phosphorylated CREB (p-CREB) protein expression was measured by Western blotting. Compared to the sham-operation group, both model and penehyclidine hydrochloride groups showed increased Zea-Longa scores. Escape latencies were longer and platform crossings were fewer in model and penehyclidine hydrochloride groups compared to the sham-operation group, but penehyclidine hydrochloride demonstrated a shorter latency and more platform crossings than the model group. BDNF and NGF content decreased in model and penehyclidine hydrochloride groups compared to the sham-operation group, with an increase in the penehyclidine hydrochloride group compared to the model group. mRNA expression levels declined in model and penehyclidine hydrochloride groups but were higher in the latter. p-CREB protein expression was lower in model and penehyclidine hydrochloride groups compared to the sham-operation group but higher in the penehyclidine hydrochloride group than the model group. Penehyclidine hydrochloride exhibited neuroprotective effects by upregulating the cAMP/CREB signaling pathway, improving cognitive function in rats with brain injury.


Assuntos
Lesões Encefálicas , Fator Neurotrófico Derivado do Encéfalo , Cognição , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , AMP Cíclico , Quinuclidinas , Ratos Sprague-Dawley , Transdução de Sinais , Animais , Transdução de Sinais/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Quinuclidinas/farmacologia , Quinuclidinas/uso terapêutico , Cognição/efeitos dos fármacos , Masculino , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , AMP Cíclico/metabolismo , Ratos , Fator de Crescimento Neural/metabolismo , Fator de Crescimento Neural/genética , Fosforilação/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Modelos Animais de Doenças
7.
Am J Chin Med ; 52(3): 799-819, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38752843

RESUMO

Subarachnoid hemorrhage (SAH), a specific subtype of cerebrovascular accident, is characterized by the extravasation of blood into the interstice between the brain and its enveloping delicate tissues. This pathophysiological phenomenon can precipitate an early brain injury (EBI), which is characterized by inflammation and neuronal death. Rutaecarpine (Rut), a flavonoid compound discovered in various plants, has been shown to have protective effects against SAH-induced cerebral insult in rodent models. In our study, we used a rodent SAH model to evaluate the effect of Rut on EBI and investigated the effect of Rut on the inflammatory response and its regulation of SIRT6 expression in vitro. We found that Rut exerts a protective effect on EBI in SAH rats, which is partly due to its ability to inhibit the inflammatory response. Notably, Rut up-regulated Sirtuin 6 (SIRT6) expression, leading to an increase in H3K9 deacetylation and inhibition of nuclear factor-kappa B (NF-[Formula: see text]B) transcriptional activation, thereby mediating the inflammatory response. In addition, further data showed that SIRT6 was proven to mediate the regulation of Rut on the microglial inflammatory response. These findings highlight the importance of SIRT6 in the regulation of inflammation and suggest a potential mechanism for the protective effect of Rut on EBI. In summary, Rut may have the potential to prevent and treat SAH-induced brain injury by interacting with SIRT6. Our findings may provide a new therapeutic strategy for the treatment of SAH-induced EBI.


Assuntos
Alcaloides Indólicos , NF-kappa B , Quinazolinas , Ratos Sprague-Dawley , Sirtuínas , Hemorragia Subaracnóidea , Animais , Hemorragia Subaracnóidea/tratamento farmacológico , Hemorragia Subaracnóidea/complicações , Sirtuínas/metabolismo , Sirtuínas/genética , Alcaloides Indólicos/farmacologia , NF-kappa B/metabolismo , Masculino , Quinazolinas/farmacologia , Quinazolinas/uso terapêutico , Modelos Animais de Doenças , Lesões Encefálicas/etiologia , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Ratos , Inflamação/tratamento farmacológico , Inflamação/etiologia , Fitoterapia , Transdução de Sinais/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Quinazolinonas
8.
Cell Rep Med ; 5(5): 101522, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38701781

RESUMO

Neuroinflammation plays a significant role in ischemic injury, which can be promoted by oxidized mitochondrial DNA (Ox-mtDNA). Cytidine/uridine monophosphate kinase 2 (CMPK2) regulates mtDNA replication, but its role in neuroinflammation and ischemic injury remains unknown. Here, we report that CMPK2 expression is upregulated in monocytes/macrophages and microglia post-stroke in humans and mice, respectively. Microglia/macrophage CMPK2 knockdown using the Cre recombination-dependent adeno-associated virus suppresses the inflammatory responses in the brain, reduces infarcts, and improves neurological outcomes in ischemic CX3CR1Cre/ERT2 mice. Mechanistically, CMPK2 knockdown limits newly synthesized mtDNA and Ox-mtDNA formation and subsequently blocks NLRP3 inflammasome activation in microglia/macrophages. Nordihydroguaiaretic acid (NDGA), as a CMPK2 inhibitor, is discovered to reduce neuroinflammation and ischemic injury in mice and prevent the inflammatory responses in primary human monocytes from ischemic patients. Thus, these findings identify CMPK2 as a promising therapeutic target for ischemic stroke and other brain disorders associated with neuroinflammation.


Assuntos
AVC Isquêmico , Microglia , Doenças Neuroinflamatórias , Animais , Humanos , Masculino , Camundongos , Lesões Encefálicas/patologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/genética , Isquemia Encefálica/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/genética , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Inflamassomos/metabolismo , AVC Isquêmico/patologia , AVC Isquêmico/metabolismo , AVC Isquêmico/genética , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/patologia , Monócitos/metabolismo , Monócitos/efeitos dos fármacos , Doenças Neuroinflamatórias/patologia , Doenças Neuroinflamatórias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética
9.
Neurobiol Dis ; 197: 106528, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38740348

RESUMO

BACKGROUND: Brain injury has been suggested as a risk factor for neurodegenerative diseases. Accordingly, defects in the brain's intrinsic capacity to repair injury may result in the accumulation of damage and a progressive loss of brain function. The G2019S (GS) mutation in LRRK2 (leucine rich repeat kinase 2) is the most prevalent genetic alteration in Parkinson's disease (PD). Here, we sought to investigate how this LRRK2-GS mutation affects repair of the injured brain. METHODS: Brain injury was induced by stereotaxic injection of ATP, a damage-associated molecular pattern (DAMP) component, into the striatum of wild-type (WT) and LRRK2-GS mice. Effects of the LRRK2-GS mutation on brain injury and the recovery from injury were examined by analyzing the molecular and cellular behavior of neurons, astrocytes, and monocytes. RESULTS: Damaged neurons express osteopontin (OPN), a factor associated with brain repair. Following ATP-induced damage, monocytes entered injured brains, phagocytosing damaged neurons and producing exosome-like vesicles (EVs) containing OPN through activation of the inflammasome and subsequent pyroptosis. Following EV production, neurons and astrocytes processes elongated towards injured cores. In LRRK2-GS mice, OPN expression and monocytic pyroptosis were decreased compared with that in WT mice, resulting in diminished release of OPN-containing EVs and attenuated elongation of neuron and astrocyte processes. In addition, exosomes prepared from injured LRRK2-GS brains induced neurite outgrowth less efficiently than those from injured WT brains. CONCLUSIONS: The LRRK2-GS mutation delays repair of injured brains through reduced expression of OPN and diminished release of OPN-containing EVs from monocytes. These findings suggest that the LRRK2-GS mutation may promote the development of PD by delaying the repair of brain injury.


Assuntos
Lesões Encefálicas , Exossomos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Monócitos , Mutação , Osteopontina , Animais , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Camundongos , Exossomos/metabolismo , Exossomos/genética , Osteopontina/metabolismo , Osteopontina/genética , Lesões Encefálicas/metabolismo , Lesões Encefálicas/genética , Monócitos/metabolismo , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/patologia , Masculino , Astrócitos/metabolismo
10.
Sci Rep ; 14(1): 10790, 2024 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-38734737

RESUMO

In this two-center prospective cohort study of children on ECMO, we assessed a panel of plasma brain injury biomarkers using exploratory factor analysis (EFA) to evaluate their interplay and association with outcomes. Biomarker concentrations were measured daily for the first 3 days of ECMO support in 95 participants. Unfavorable composite outcome was defined as in-hospital mortality or discharge Pediatric Cerebral Performance Category > 2 with decline ≥ 1 point from baseline. EFA grouped 11 biomarkers into three factors. Factor 1 comprised markers of cellular brain injury (NSE, BDNF, GFAP, S100ß, MCP1, VILIP-1, neurogranin); Factor 2 comprised markers related to vascular processes (vWF, PDGFRß, NPTX1); and Factor 3 comprised the BDNF/MMP-9 cellular pathway. Multivariable logistic models demonstrated that higher Factor 1 and 2 scores were associated with higher odds of unfavorable outcome (adjusted OR 2.88 [1.61, 5.66] and 1.89 [1.12, 3.43], respectively). Conversely, higher Factor 3 scores were associated with lower odds of unfavorable outcome (adjusted OR 0.54 [0.31, 0.88]), which is biologically plausible given the role of BDNF in neuroplasticity. Application of EFA on plasma brain injury biomarkers in children on ECMO yielded grouping of biomarkers into three factors that were significantly associated with unfavorable outcome, suggesting future potential as prognostic instruments.


Assuntos
Biomarcadores , Lesões Encefálicas , Oxigenação por Membrana Extracorpórea , Humanos , Biomarcadores/sangue , Masculino , Feminino , Recém-Nascido , Lactente , Lesões Encefálicas/sangue , Lesões Encefálicas/terapia , Lesões Encefálicas/diagnóstico , Lesões Encefálicas/metabolismo , Criança , Pré-Escolar , Estudos Prospectivos , Análise Fatorial , Mortalidade Hospitalar , Resultado do Tratamento
11.
Free Radic Biol Med ; 220: 271-287, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38734267

RESUMO

Bilirubin-induced brain damage is a serious clinical consequence of hyperbilirubinemia, yet the underlying molecular mechanisms remain largely unknown. Ferroptosis, an iron-dependent cell death, is characterized by iron overload and lipid peroxidation. Here, we report a novel regulatory mechanism of demethylase AlkB homolog 5 (ALKBH5) in acyl-coenzyme A synthetase long-chain family member 4 (ACSL4)-mediated ferroptosis in hyperbilirubinemia. Hyperdifferential PC12 cells and newborn Sprague-Dawley rats were used to establish in vitro and in vivo hyperbilirubinemia models, respectively. Proteomics, coupled with bioinformatics analysis, first suggested the important role of ferroptosis in hyperbilirubinemia-induced brain damage. In vitro experiments showed that ferroptosis is activated in hyperbilirubinemia, and ferroptosis inhibitors (desferrioxamine and ferrostatin-1) treatment effectively alleviates hyperbilirubinemia-induced oxidative damage. Notably, we observed that the ferroptosis in hyperbilirubinemia was regulated by m6A modification through the downregulation of ALKBH5 expression. MeRIP-seq and RIP-seq showed that ALKBH5 may trigger hyperbilirubinemia ferroptosis by stabilizing ACSL4 mRNA via m6A modification. Further, hyperbilirubinemia-induced oxidative damage was alleviated through ACSL4 genetic knockdown or rosiglitazone-mediated chemical repression but was exacerbated by ACSL4 overexpression. Mechanistically, ALKBH5 promotes ACSL4 mRNA stability and ferroptosis by combining the 669 and 2015 m6A modified sites within 3' UTR of ACSL4 mRNA. Our findings unveil a novel molecular mechanism of ferroptosis and suggest that m6A-dependent ferroptosis could be an underlying clinical target for the therapy of hyperbilirubinemia.


Assuntos
Homólogo AlkB 5 da RNA Desmetilase , Coenzima A Ligases , Ferroptose , Estabilidade de RNA , Ratos Sprague-Dawley , Animais , Ferroptose/genética , Ratos , Coenzima A Ligases/genética , Coenzima A Ligases/metabolismo , Homólogo AlkB 5 da RNA Desmetilase/metabolismo , Homólogo AlkB 5 da RNA Desmetilase/genética , Células PC12 , Cicloexilaminas/farmacologia , Humanos , Desferroxamina/farmacologia , Estresse Oxidativo , Lesões Encefálicas/metabolismo , Lesões Encefálicas/genética , Lesões Encefálicas/patologia , Lesões Encefálicas/etiologia , Fenilenodiaminas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Masculino , Modelos Animais de Doenças , Peroxidação de Lipídeos
12.
ACS Nano ; 18(21): 13836-13848, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38753820

RESUMO

Advanced in vivo imaging techniques have facilitated the comprehensive visual exploration of animal biological processes, leading to groundbreaking discoveries such as the glymphatic system. However, current limitations of macroscopic imaging techniques impede the precise investigation of physiological parameters regulating this specialized lymphatic transport system. While NIR-II fluorescence imaging has demonstrated advantages in peripheral lymphatic imaging, there are few reports regarding its utilization in the glymphatic system. To address this, a noninvasive transcranial macroscopic NIR-II fluorescence imaging model is developed using a cyanine dye-protein coupled nanoprobe. NIR-II imaging with high temporal and spatial resolution reveals that hypothermia can increase the glymphatic influx by reducing the flow rate of cerebrospinal fluid. In addition, respiratory rate, respiratory amplitude, and heart rate all play a role in regulating the glymphatic influx. Thus, targeting the glymphatic influx may alter the trajectory of immune inflammation following brain injury, providing therapeutic prospects for treating brain injury with mild hypothermia.


Assuntos
Lesões Encefálicas , Sistema Glinfático , Animais , Sistema Glinfático/diagnóstico por imagem , Sistema Glinfático/metabolismo , Lesões Encefálicas/metabolismo , Lesões Encefálicas/diagnóstico por imagem , Lesões Encefálicas/terapia , Camundongos , Imagem Óptica , Hipotermia/metabolismo , Doenças Neuroinflamatórias/diagnóstico por imagem , Doenças Neuroinflamatórias/metabolismo , Raios Infravermelhos , Corantes Fluorescentes/química , Masculino , Hipotermia Induzida , Camundongos Endogâmicos C57BL , Carbocianinas/química
13.
Proc Natl Acad Sci U S A ; 121(22): e2400648121, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38781210

RESUMO

After central nervous system injury, a rapid cellular and molecular response is induced. This response can be both beneficial and detrimental to neuronal survival in the first few days and increases the risk for neurodegeneration if persistent. Semaphorin4B (Sema4B), a transmembrane protein primarily expressed by cortical astrocytes, has been shown to play a role in neuronal cell death following injury. Our study shows that after cortical stab wound injury, cytokine expression is attenuated in Sema4B-/- mice, and microglia/macrophage reactivity is altered. In vitro, Sema4B enhances the reactivity of microglia following injury, suggesting astrocytic Sema4B functions as a ligand. Moreover, injury-induced microglia reactivity is attenuated in the presence of Sema4B-/- astrocytes compared to Sema4B+/- astrocytes. In vitro experiments indicate that Plexin-B2 is the Sema4B receptor on microglia. Consistent with this, in microglia/macrophage-specific Plexin-B2-/- mice, similar to Sema4B-/- mice, microglial/macrophage reactivity and neuronal cell death are attenuated after cortical injury. Finally, in Sema4B/Plexin-B2 double heterozygous mice, microglial/macrophage reactivity is also reduced after injury, supporting the idea that both Sema4B and Plexin-B2 are part of the same signaling pathway. Taken together, we propose a model in which following injury, astrocytic Sema4B enhances the response of microglia/macrophages via Plexin-B2, leading to increased reactivity.


Assuntos
Astrócitos , Camundongos Knockout , Microglia , Proteínas do Tecido Nervoso , Semaforinas , Animais , Camundongos , Astrócitos/metabolismo , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Lesões Encefálicas/genética , Comunicação Celular , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/patologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Semaforinas/metabolismo , Semaforinas/genética
14.
Int Immunopharmacol ; 135: 112311, 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38781607

RESUMO

OBJECTIVE: Obstructive Sleep Apnea (OSA) during pregnancy is characterized by intermittent hypoxia (IH) during sleep and will lead to the rise of oxidative stress in the fetal body. Pyroptosis, a type of inflammatory and programmable cell death mediated by Gasdermin D (GSDMD), plays a substantial role in oxygen deprivation's contribution to neural system damage. Existing research shows that Nicotinamide Adenine Dinucleotide Phosphate (NADPH) plays a protective role in alleviating brain tissue pyroptosis. We speculate that exogenous NADPH may play a protective role in OSA during pregnancy. METHODS: A model of GIH group was established to simulate the pathophysiological mechanisms of OSA during pregnant and AIR group was established by giving the same frequency. Sham group was established by injecting NS and the NADPH group was established and given exogenous NADPH. We utilized the Morris Water Maze to assess cognitive function impairment, Luxol Fast Blue (LBF) staining to confirm myelin sheath formation, TUNEL staining to examine cell death in fetal mice brain tissue, and Western blotting to detect pertinent protein expressions. RESULTS: The GIH group offspring exhibited decreases in spatial learning and memory abilities, reduced numbers of oligodendrocytes and formed myelin, as well as increased expression of pyroptosis-related proteins. The NADPH group offspring showed restoration in spatial learning and memory abilities increased counts of oligodendrocytes and formed myelin sheaths, in addition to decreased expression of pyroptosis-related. CONCLUSIONS: This study demonstrates that early injection of exogenous NADPH can alleviate the damage to fetal brain development caused by gestational intermittent hypoxia (GIH).


Assuntos
NADP , Piroptose , Animais , Gravidez , Feminino , Camundongos , NADP/metabolismo , Lesões Encefálicas/patologia , Lesões Encefálicas/metabolismo , Hipóxia/metabolismo , Encéfalo/patologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL , Efeitos Tardios da Exposição Pré-Natal
16.
EMBO Mol Med ; 16(6): 1228-1253, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38789599

RESUMO

In the injured brain, new neurons produced from endogenous neural stem cells form chains and migrate to injured areas and contribute to the regeneration of lost neurons. However, this endogenous regenerative capacity of the brain has not yet been leveraged for the treatment of brain injury. Here, we show that in healthy brain chains of migrating new neurons maintain unexpectedly large non-adherent areas between neighboring cells, allowing for efficient migration. In instances of brain injury, neuraminidase reduces polysialic acid levels, which negatively regulates adhesion, leading to increased cell-cell adhesion and reduced migration efficiency. The administration of zanamivir, a neuraminidase inhibitor used for influenza treatment, promotes neuronal migration toward damaged regions, fosters neuronal regeneration, and facilitates functional recovery. Together, these findings shed light on a new mechanism governing efficient neuronal migration in the adult brain under physiological conditions, pinpoint the disruption of this mechanism during brain injury, and propose a promising therapeutic avenue for brain injury through drug repositioning.


Assuntos
Encéfalo , Movimento Celular , Neuraminidase , Neurônios , Neuraminidase/metabolismo , Neuraminidase/antagonistas & inibidores , Movimento Celular/efeitos dos fármacos , Animais , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Camundongos , Zanamivir/farmacologia , Inibidores Enzimáticos/farmacologia , Ácidos Siálicos/metabolismo , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Recuperação de Função Fisiológica/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Adesão Celular/efeitos dos fármacos , Humanos , Masculino
17.
Front Immunol ; 15: 1386780, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38756773

RESUMO

Introduction: Intracerebral hemorrhage (ICH) often triggers oxidative stress through reactive oxygen species (ROS). Transforming growth factor-ß-activated kinase 1 (TAK1) plays a pivotal role in regulating oxidative stress and inflammation across various diseases. 5Z-7-Oxozeaenol (OZ), a specific inhibitor of TAK1, has exhibited therapeutic effects in various conditions. However, the impact of OZ following ICH and its underlying molecular mechanisms remain elusive. This study aimed to explore the possible role of OZ in ICH and its underlying mechanisms by inhibiting oxidative stress-mediated pyroptosis. Methods: Adult male Sprague-Dawley rats were subjected to an ICH model, followed by treatment with OZ. Neurobehavioral function, blood-brain barrier integrity, neuronal pyroptosis, and oxidative stress markers were assessed using various techniques including behavioral tests, immunofluorescence staining, western blotting, transmission electron microscopy, and biochemical assays. Results: Our study revealed that OZ administration significantly inhibited phosphorylated TAK1 expression post-ICH. Furthermore, TAK1 blockade by OZ attenuated blood-brain barrier (BBB) disruption, neuroinflammation, and oxidative damage while enhancing neurobehavioral function. Mechanistically, OZ administration markedly reduced ROS production and oxidative stress by facilitating nuclear factor-erythroid 2-related factor 2 (NRF2) nuclear translocation. This was accompanied by a subsequent suppression of the NOD-like receptor protein 3 (NLRP3) activation-mediated inflammatory cascade and neuronal pyroptosis. Discussion: Our findings highlight that OZ alleviates brain injury and oxidative stress-mediated pyroptosis via the NRF2 pathway. Inhibition of TAK1 emerges as a promising approach for managing ICH.


Assuntos
Hemorragia Cerebral , MAP Quinase Quinase Quinases , Fator 2 Relacionado a NF-E2 , Neurônios , Estresse Oxidativo , Piroptose , Ratos Sprague-Dawley , Transdução de Sinais , Animais , Piroptose/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/tratamento farmacológico , Masculino , Ratos , Transdução de Sinais/efeitos dos fármacos , MAP Quinase Quinase Quinases/metabolismo , MAP Quinase Quinase Quinases/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Modelos Animais de Doenças , Lesões Encefálicas/etiologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/tratamento farmacológico , Espécies Reativas de Oxigênio/metabolismo , Lactonas , Resorcinóis , Zearalenona/administração & dosagem
18.
J Neuroinflammation ; 21(1): 116, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38702778

RESUMO

BACKGROUND: Subarachnoid hemorrhage (SAH), a severe subtype of stroke, is characterized by notably high mortality and morbidity, largely due to the lack of effective therapeutic options. Although the neuroprotective potential of PPARg and Nrf2 has been recognized, investigative efforts into oroxin A (OA), remain limited in preclinical studies. METHODS: SAH was modeled in vivo through filament perforation in male C57BL/6 mice and in vitro by exposing HT22 cells to hemin to induce neuronal damage. Following the administration of OA, a series of methods were employed to assess neurological behaviors, brain water content, neuronal damage, cell ferroptosis, and the extent of neuroinflammation. RESULTS: The findings indicated that OA treatment markedly improved survival rates, enhanced neurological functions, mitigated neuronal death and brain edema, and attenuated the inflammatory response. These effects of OA were linked to the suppression of microglial activation. Moreover, OA administration was found to diminish ferroptosis in neuronal cells, a critical factor in early brain injury (EBI) following SAH. Further mechanistic investigations uncovered that OA facilitated the translocation of nuclear factor erythroid 2-related factor 2 (Nrf-2) from the cytoplasm to the nucleus, thereby activating the Nrf2/GPX4 pathway. Importantly, OA also upregulated the expression of FSP1, suggesting a significant and parallel protective effect against ferroptosis in EBI following SAH in synergy with GPX4. CONCLUSION: In summary, this research indicated that the PPARg activator OA augmented the neurological results in rodent models and diminished neuronal death. This neuroprotection was achieved primarily by suppressing neuronal ferroptosis. The underlying mechanism was associated with the alleviation of cellular death through the Nrf2/GPX4 and FSP1/CoQ10 pathways.


Assuntos
Ferroptose , Camundongos Endogâmicos C57BL , Doenças Neuroinflamatórias , Hemorragia Subaracnóidea , Animais , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/patologia , Hemorragia Subaracnóidea/complicações , Ferroptose/efeitos dos fármacos , Ferroptose/fisiologia , Camundongos , Masculino , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/etiologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Lesões Encefálicas/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia
19.
J Neurosci Res ; 102(5): e25356, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38773875

RESUMO

From the blood brain barrier to the synaptic space, astrocytes provide structural, metabolic, ionic, and extracellular matrix (ECM) support across the brain. Astrocytes include a vast array of subtypes, their phenotypes and functions varying both regionally and temporally. Astrocytes' metabolic and regulatory functions poise them to be quick and sensitive responders to injury and disease in the brain as revealed by single cell sequencing. Far less is known about the influence of the local healthy and aging microenvironments on these astrocyte activation states. In this forward-looking review, we describe the known relationship between astrocytes and their local microenvironment, the remodeling of the microenvironment during disease and injury, and postulate how they may drive astrocyte activation. We suggest technology development to better understand the dynamic diversity of astrocyte activation states, and how basal and activation states depend on the ECM microenvironment. A deeper understanding of astrocyte response to stimuli in ECM-specific contexts (brain region, age, and sex of individual), paves the way to revolutionize how the field considers astrocyte-ECM interactions in brain injury and disease and opens routes to return astrocytes to a healthy quiescent state.


Assuntos
Astrócitos , Encéfalo , Matriz Extracelular , Astrócitos/fisiologia , Astrócitos/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiologia , Humanos , Animais , Encéfalo/metabolismo , Lesões Encefálicas/patologia , Lesões Encefálicas/metabolismo
20.
Brain Res ; 1836: 148909, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38570154

RESUMO

BACKGROUND: Early brain injury (EBI) is closely associated with poor prognosis in patients with subarachnoid haemorrhage (SAH), with autophagy playing a pivotal role in EBI. However, research has shown that the stimulator of interferon genes (STING) pathway impacts autophagic flux. While the regulatory impact of neuritin on EBI and autophagic flux has been established previously, the underlying mechanism remains unclear. This study aimed to determine the role of the cGAS-STING pathway in neuritin-mediated regulation of autophagic flux following SAH. METHODS: A SAH model was established in male Sprague-Dawley rats via intravascular perforation. Neuritin overexpressions using adeno-associated virus, the STING antagonist "C-176," and the activator, "CMA," were determined to investigate the cGAS-STING pathway's influence on autophagic flux and brain injury post-SAH, along with the neuritin's regulatory effect on STING. In this study, SAH grade, neurological score, haematoxylin and eosin (H&E) staining, brain water content (BWC), sandwich enzyme-linked immunosorbent assay, Evans blue staining, immunofluorescence staining, western blot analysis, and transmission electron microscopy (TEM) were examined. RESULTS: Neuritin overexpression significantly ameliorated neurobehavioural scores, blood-brain barrier injury, brain oedema, and impaired autophagic flux in SAH-induced rats. STING expression remarkably increased post-SAH. C-176 and CMA mitigated and aggravated autophagic flux injury and brain injury, respectively, while inhibiting and enhancing STING, respectively. Particularly, CMA treatment nullified the protective effects of neuritin against autophagic flux and mitigated brain injury. CONCLUSION: Neuritin alleviated EBI by restoring impaired autophagic flux after SAH through the regulation of the cGAS-STING pathway.


Assuntos
Autofagia , Lesões Encefálicas , Proteínas de Membrana , Ratos Sprague-Dawley , Transdução de Sinais , Hemorragia Subaracnóidea , Animais , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Masculino , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/complicações , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Lesões Encefálicas/metabolismo , Proteínas de Membrana/metabolismo , Neuropeptídeos/metabolismo , Proteínas Ligadas por GPI/metabolismo , Modelos Animais de Doenças
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA